• Nenhum resultado encontrado

Este estudo teve como hipótese que a expressão gênica para ADAM10 e miRNAs em amostras de sangue encontram-se alteradas em idosos com DA em comparação a idosos sem alteração cognitiva.

Sendo assim, os objetivos gerais deste projeto foram:

1) Verificar e comparar a expressão gênica da ADAM10 em amostras de sangue entre sujeitos com TNCL, DA e sem alteração cognitiva;

2) Verificar e comparar a expressão gênica de miRNAs em amostras de sangue entre sujeitos com DA e sem alteração cognitiva;

3) Verificar se existe relação entre os níveis de mRNA para a ADAM10 e a expressão de miRNAs, visando fortalecer o papel da ADAM10 como molécula biomarcadora para a DA.

REFERÊNCIAS

BIBLIOGRÁFICAS

Referências Bibliográficas

3. REFERÊNCIAS BIBLIOGRÁFICAS

ALBERT, M. S., et al. (2011). The diagnosis of mild cognitive impairment due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers

Dement, 7, 270-279.

AMBROS, V. (2004). The functions of animal microRNAs. Nature, 431, 350-355.

AMBROS, V. (2010). MicroRNAs: genetically sensitized worms reveal new secrets. Curr

Biol, 20, R598-600.

ANDERS, A., GILBERT, S., GARTEN, W., POSTINA, R. AND FAHRENHOLZ, F. (2001). Regulation of the alpha-secretase ADAM10 by its prodomain and proprotein convertases.

Faseb J, 15, 1837-1839.

ANDERSSON, E. R., SANDBERG, R. AND LENDAHL, U. (2011). Notch signaling: simplicity in design, versatility in function. Development, 138, 3593-3612.

AUGUSTIN, R., et al. (2012). Computational identification and experimental validation of microRNAs binding to the Alzheimer-related gene ADAM10. BMC Med Genet, 13, 35. BALLARD, C., GAUTHIER, S., CORBETT, A., BRAYNE, C., AARSLAND, D. AND

JONES, E. (2011). Alzheimer's disease. Lancet, 377, 1019-1031.

BANDYOPADHYAY, S., HARTLEY, D. M., CAHILL, C. M., LAHIRI, D. K., CHATTOPADHYAY, N. AND ROGERS, J. T. (2006). Interleukin-1 stimulates non- amyloidogenic pathway by -secretase (ADAM-10 and ADAM-17) cleavage of APP in human astrocytic cells involving p38 MAP kinase. Journal of Neuroscience Research, 84, 106-118.

BARAO, S., et al. (2013). BACE1 levels correlate with phospho-tau levels in human cerebrospinal fluid. Current Alzheimer Research, 10, 671-678.

BEREZIKOV, E., et al. (2006). Diversity of microRNAs in human and chimpanzee brain.

Nat Genet, 38, 1375-1377.

BLASKO, I. J., K.; KEMMLER, G. (2008). Conversion from cognitive health to mild cognitive impairment and Alzheimer's disease: Prediction by plasma amyloid beta 42, medial temporal lobe atrophy and homocysteine. Neurobiology of aging, 29, 1-11.

BOISSONNEAULT, V., PLANTE, I., RIVEST, S. AND PROVOST, P. (2009). MicroRNA- 298 and microRNA-328 regulate expression of mouse beta-amyloid precursor protein- converting enzyme 1. J Biol Chem, 284, 1971-1981.

CHAIMOWICZ, F. (1997). [Health of Brazilian elderly just before of the 21st century: current problems, forecasts and alternatives]. Rev Saude Publica, 31, 184-200.

Referências Bibliográficas

CHANTRY, A., GREGSON, N. A. AND GLYNN, P. (1989). A novel metalloproteinase associated with brain myelin membranes. Isolation and characterization. J Biol Chem, 264, 21603-21607.

COGSWELL, J. P., et al. (2008). Identification of miRNA changes in Alzheimer's disease brain and CSF yields putative biomarkers and insights into disease pathways. J Alzheimers

Dis, 14, 27-41.

COLCIAGHI, F., et al. (2002). [alpha]-Secretase ADAM10 as well as [alpha]APPs is reduced in platelets and CSF of Alzheimer disease patients. Mol Med, 8, 67-74.

COLCIAGHI, F., et al. (2004). Platelet APP, ADAM 10 and BACE alterations in the early stages of Alzheimer disease. Neurology, 62, 498-501.

COLE, S. L. AND VASSAR, R. (2007). The Alzheimer's disease beta-secretase enzyme, BACE1. Mol Neurodegener, 2, 22.

DE BARRY, J., LIEGEOIS, C. M. AND JANOSHAZI, A. (2010). Protein kinase C as a peripheral biomarker for Alzheimer's disease. Exp Gerontol, 45, 64-69.

DE SMAELE, E., FERRETTI, E. AND GULINO, A. (2010). MicroRNAs as biomarkers for CNS cancer and other disorders. Brain Research, 1338, 100-111.

DELAY, C., MANDEMAKERS, W. AND HEBERT, S. S. (2012). MicroRNAs in Alzheimer's disease. Neurobiology of Disease, 46, 285-290.

DENG, W., CHO, S., SU, P. C., BERGER, B. W. AND LI, R. (2014). Membrane-enabled dimerization of the intrinsically disordered cytoplasmic domain of ADAM10. Proc Natl

Acad Sci U S A, 111, 15987-15992.

DI LUCA, M., COLCIAGHI, F., PASTORINO, L., BORRONI, B., PADOVANI, A. AND CATTABENI, F. (2000). Platelets as a peripheral district where to study pathogenetic mechanisms of Alzheimer disease: the case of amyloid precursor protein. European

Journal of Pharmacology, 405, 277-283.

DONMEZ, G., WANG, D., COHEN, D. E. AND GUARENTE, L. (2010). SIRT1 Suppresses ²-Amyloid Production by Activating the ±-Secretase Gene ADAM10. Cell, 142, 320-332. DSM-V (2013). Diagnostic and statistical manual of mental disorders. 5th ed. American

Psychiatric Association

EDISON, P., et al. (2007). Amyloid, hypometabolism, and cognition in Alzheimer disease: an [11C]PIB and [18F]FDG PET study. Neurology, 68, 501-508.

ENDRES, K., et al. (2014). Increased CSF APPs-alpha levels in patients with Alzheimer disease treated with acitretin. Neurology, 83, 1930-1935.

ENDRES, K., POSTINA, R., SCHROEDER, A., MUELLER, U. AND FAHRENHOLZ, F. (2005). Shedding of the amyloid precursor protein-like protein APLP2 by disintegrin- metalloproteinases. Febs J, 272, 5808-5820.

Referências Bibliográficas

ESQUELA-KERSCHER, A. AND SLACK, F. J. (2006). Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer, 6, 259-269.

EVIN, G., ZHU, A., HOLSINGER, R. M. D., MASTERS, C. L. AND LI, Q.-X. (2003). Proteolytic processing of the Alzheimer's disease amyloid precursor protein in brain and platelets. Journal of Neuroscience Research, 74, 386-392.

FAHRENHOLZ, F. (2010). The close link between retinoid signalling and the -secretase ADAM10 and its potential for treating Alzheimer’s disease (Commentary on Jarvis et al.).

European Journal of Neuroscience, 32, 1245-1245.

FOX, J. W. AND SERRANO, S. M. (2005). Structural considerations of the snake venom metalloproteinases, key members of the M12 reprolysin family of metalloproteinases.

Toxicon, 45, 969-985.

GHOSAL, K., VOGT, D. L., LIANG, M., SHEN, Y., LAMB, B. T. AND PIMPLIKAR, S. W. (2009). Alzheimer's disease-like pathological features in transgenic mice expressing the APP intracellular domain. Proc Natl Acad Sci U S A, 106, 18367-18372.

GRAFF-RADFORD, N. R. C., J.E.; LUCAS, et al. (2007). Association of low plasma A 42/A 40 ratios with increased imminent risk for mild cognitive impairment and Alzheimer disease. Arch. Neurol, 64.

GUTWEIN, P., et al. (2003). ADAM10-mediated cleavage of L1 adhesion molecule at the cell surface and in released membrane vesicles. Faseb J, 17, 292-294.

HERRERA, E. C., P.; NITRINI, R. (1998). Estudo epidemiológico populacional de demência na cidade de Catanduva, Estado de São Paulo, Brasil. . Rev. Psiquatr. Clin, 25, 70-73. HOLSINGER, R. M., MCLEAN, C. A., BEYREUTHER, K., MASTERS, C. L. AND EVIN,

G. (2002). Increased expression of the amyloid precursor beta-secretase in Alzheimer's disease. Ann Neurol, 51, 783-786.

HOOPER, N. M. AND TURNER, A. J. (2002). The search for alpha-secretase and its potential as a therapeutic approach to Alzheimer s disease. Curr Med Chem, 9, 1107-1119. HOWARD, L., LU, X., MITCHELL, S., GRIFFITHS, S. AND GLYNN, P. (1996).

Molecular cloning of MADM: a catalytically active mammalian disintegrin- metalloprotease expressed in various cell types. Biochemical Journal, 317, 45-50.

HWANG, E. M., et al. (2006). Furin is an endogenous regulator of alpha-secretase associated APP processing. Biochem Biophys Res Commun, 349, 654-659.

IBGE (2010). Estimativa da população idosa brasileira.

JALBERT, J. J., DAIELLO, L. A. AND LAPANE, K. L. (2008). Dementia of the Alzheimer type. Epidemiol rev, 30, 15-34.

JORISSEN, E., et al. (2010). The Disintegrin/Metalloproteinase ADAM10 Is Essential for the Establishment of the Brain Cortex. Journal of Neuroscience, 30, 4833-4844.

Referências Bibliográficas

KARKKAINEN, I., RYBNIKOVA, E., PELTO-HUIKKO, M. AND HUOVILA, A. P. (2000). Metalloprotease-disintegrin (ADAM) genes are widely and differentially expressed in the adult CNS. Mol Cell Neurosci, 15, 547-560.

KELLER, A., et al. (2011). Toward the blood-borne miRNome of human diseases. Nat Meth,

8, 841-843.

KIKO, T., NAKAGAWA, K., TSUDUKI, T., FURUKAWA, K., ARAI, H. AND MIYAZAWA, T. (2014). MicroRNAs in plasma and cerebrospinal fluid as potential markers for Alzheimer's disease. J Alzheimers Dis, 39, 253-259.

KIM, M., et al. (2009). Potential late-onset Alzheimer's disease-associated mutations in the ADAM10 gene attenuate {alpha}-secretase activity. Hum Mol Genet, 18, 3987-3996. KOJRO, E., GIMPL, G., LAMMICH, S., MÄRZ, W. AND FAHRENHOLZ, F. (2001). Low

cholesterol stimulates the nonamyloidogenic pathway by its effect on the -secretase ADAM 10. Proceedings of the National Academy of Sciences of the United States of

America, 98, 5815-5820.

KROL, J., LOEDIGE, I. AND FILIPOWICZ, W. (2010). The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet, 11, 597-610.

KUHN, P. H., et al. (2010). ADAM10 is the physiologically relevant, constitutive alpha- secretase of the amyloid precursor protein in primary neurons. Embo Journal, 29, 3020- 3032.

KUHN, P.H., COLOMBO, A.V., SCHUSSER, B. et al. (2016). Systematic substrate identification indicates a central role for the metalloprotease ADAM10 in axon targeting and synapse function. eLife, 23, 10.7554/eLife.12748.

KUMAR, P., et al. (2013). Circulating miRNA biomarkers for Alzheimer's disease. PLoS

One, 8.

LAMMICH, S., et al. (2010). Expression of the anti-amyloidogenic secretase ADAM10 is suppressed by its 5'-untranslated region. J Biol Chem, 285, 15753-15760.

LAMMICH, S., et al. (1999). Constitutive and regulated alpha-secretase cleavage of Alzheimer's amyloid precursor protein by a disintegrin metalloprotease. Proc Natl Acad

Sci U S A, 96, 3922-3927.

LEIDINGER, P., et al. (2013). A blood based 12-miRNA signature of Alzheimer disease patients. Genome Biol, 14, 2013-2014.

LESKO, L. J. AND ATKINSON, A. J., JR. (2001). Use of biomarkers and surrogate endpoints in drug development and regulatory decision making: criteria, validation, strategies. Annu Rev Pharmacol Toxicol, 41, 347-366.

LI, Y., et al. (2008). Regional analysis of FDG and PIB-PET images in normal aging, mild cognitive impairment, and Alzheimer’s disease. European Journal of Nuclear Medicine

Referências Bibliográficas

LICHTENTHALER, S. F. (2011). Alpha-secretase in Alzheimer’s disease: molecular identity, regulation and therapeutic potential. Journal of Neurochemistry, 116, 10-21. LIN, J., LUO, J. AND REDIES, C. (2008). Differential expression of five members of the

ADAM family in the developing chicken brain. Neuroscience, 157, 360-375.

LOPEZ-PEREZ, E., ZHANG, Y., FRANK, S. J., CREEMERS, J., SEIDAH, N. AND CHECLER, F. (2001). Constitutive -secretase cleavage of the -amyloid precursor protein in the furin-deficient LoVo cell line: involvement of the pro-hormone convertase 7 and the disintegrin metalloprotease ADAM10. Journal of Neurochemistry, 76, 1532-1539. LUKIW, W. J. (2007). Micro-RNA speciation in fetal, adult and Alzheimer's disease

hippocampus. Neuroreport, 18, 297-300.

LUNN, C. A., et al. (1997). Purification of ADAM 10 from bovine spleen as a TNFalpha convertase. FEBS Lett, 400, 333-335.

MAES, O. C., XU, S., YU, B., CHERTKOW, H. M., WANG, E. AND SCHIPPER, H. M. (2007). Transcriptional profiling of Alzheimer blood mononuclear cells by microarray.

Neurobiology of aging, 28, 1795-1809.

MANZINE, P. R., BARHAM, E. J., VALE, F. A., SELISTRE-DE-ARAUJO, H. S., PAVARINI, S. C. AND COMINETTI, M. R. (2013a). Platelet a disintegrin and metallopeptidase 10 expression correlates with clock drawing test scores in Alzheimer's disease. Int J Geriatr Psychiatry, 22, 414-420.

MANZINE, P. R., BARHAM, E. J., VALE, F. A. C., SELISTRE-DE-ARAUJO, H. S., IOST PAVARINI, S. C. AND COMINETTI, M. R. (2013b). Correlation Between Mini-Mental State Examination and Platelet ADAM10 Expression in Alzheimer's Disease. J Alzheimers

Dis, 36, 253-260.

MANZINE, P. R., et al. (2013c). ADAM10 as a biomarker for Alzheimer's disease: a study with Brazilian elderly. Dement Geriatr Cogn Disord, 35, 58-66.

MAO, X., et al. The effects of chronic copper exposure on the amyloid protein metabolisim associated genes’ expression in chronic cerebral hypoperfused rats. Neuroscience Letters. MARCELLO, E., et al. (2013). Endocytosis of synaptic ADAM10 in neuronal plasticity and

Alzheimer’s disease. The Journal of Clinical Investigation, 123, 2523-2538.

MARCELLO, E., et al. (2012). SAP97-mediated local trafficking is altered in Alzheimer disease patients' hippocampus. Neurobiol Aging, 33, 27.

MARCELLO, E., GARDONI, F., DI LUCA, M. AND PEREZ-OTANO, I. (2010). An arginine stretch limits ADAM10 exit from the endoplasmic reticulum. J Biol Chem, 285, 10376-10384.

Referências Bibliográficas

MARCINKIEWICZ, M. AND SEIDAH, N. G. (2000). Coordinated expression of beta- amyloid precursor protein and the putative beta-secretase BACE and alpha-secretase ADAM10 in mouse and human brain. J Neurochem, 75, 2133-2143.

MEGURO, K., CHUBACI, R.Y.S., MEGURO, M., KAWAMORIDA, K., GOTO, N., CARAMELLI, P. (2011). Incidence of dementia and cause of death in elderly Japanese emigrants to Brazil before World War II. Arch Gerontol Geriatr, 52, 75-78.

MEGURO, K., MEGURO, M., CARAMELLI, P., ISHIZAKI, J., AMBO, H., CHUBACI, R.Y., HAMADA, G.S., NITRINI, R., YAMADORI, A. (2001). Elderly Japanese emigrants to Brazil before World War II: II. Prevalence of senile dementia. Int J Geriatr

Psychiatry, 16, 775-779.

MORISHIMA-KAWASHIMA, M. AND IHARA, Y. (2002). Alzheimer's disease: -Amyloid protein and tau. Journal of Neuroscience Research, 70, 392-401.

MOSS, M. L., et al. (2011). ADAM9 inhibition increases membrane activity of ADAM10 and controls alpha-secretase processing of amyloid precursor protein. J Biol Chem, 286, 40443-40451.

NIKOLAEV, A., MCLAUGHLIN, T., O'LEARY, D. D. AND TESSIER-LAVIGNE, M. (2009). APP binds DR6 to trigger axon pruning and neuron death via distinct caspases.

Nature, 457, 981-989.

NITRINI, R., et al. (2009). Prevalence of dementia in Latin America: a collaborative study of population-based cohorts. Int Psychogeriatr, 21, 622-630.

PAPADOPOULOS, G. L., ALEXIOU, P., MARAGKAKIS, M., RECZKO, M. AND HATZIGEORGIOU, A. G. (2009). DIANA-mirPath: Integrating human and mouse microRNAs in pathways. Bioinformatics, 25, 1991-1993.

PASCIUTO, E., et al. (2015). Dysregulated ADAM10-Mediated Processing of APP during a Critical Time Window Leads to Synaptic Deficits in Fragile X Syndrome. Neuron, 87, 382-398.

PETERSEN, R. C., SMITH, G. E., WARING, S. C., et al. (1999). Mild cognitive impairment: clinical characterization and outcome. Arch Neurol, 56, 303-308.

PETERSEN, R. C., STEVENS, J. C., GANGULI, M., TANGALOS, E. G., CUMMINGS, J. L. AND DEKOSKY, S. T. (2001). Practice parameter: early detection of dementia: mild cognitive impairment (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology, 56, 1133-1142.

PIMPLIKAR, S. W., NIXON, R. A., ROBAKIS, N. K., SHEN, J. AND TSAI, L. H. (2010). Amyloid-independent mechanisms in Alzheimer's disease pathogenesis. J Neurosci, 30, 14946-14954.

Referências Bibliográficas

PLASSMAN, B. L., et al. (2008). Prevalence of cognitive impairment without dementia in the United States. Ann Intern Med, 148, 427-434.

PRINZEN, C., TRUMBACH, D., WURST, W., et al. (2009). Differential gene expression in ADAM10 and mutant ADAM10 transgenic mice. BMC Genomics, 10, 66.

QIN, W., et al. (2009). S100A7, a novel Alzheimer's disease biomarker with non- amyloidogenic alpha-secretase activity acts via selective promotion of ADAM-10. PLoS

One, 4.

REISS, K., CORNELSEN, I., HUSMANN, M., GIMPL, G. AND BHAKDI, S. (2011). Unsaturated fatty acids drive disintegrin and metalloproteinase (ADAM)-dependent cell adhesion, proliferation, and migration by modulating membrane fluidity. J Biol Chem, 286, 26931-26942.

REISS, K. AND SAFTIG, P. (2009). The "a disintegrin and metalloprotease" (ADAM) family of sheddases: physiological and cellular functions. Semin Cell Dev Biol, 20, 126-137. SAFTIG, P. AND LICHTENTHALER, S. F. (2015). The alpha secretase ADAM10: A

metalloprotease with multiple functions in the brain. Prog Neurobiol, 29, 30046-30040. SATOH, J. (2012). Molecular network of microRNA targets in Alzheimer's disease brains.

Exp Neurol, 235, 436-446.

SAYKIN, A. J., et al. (2010). Alzheimer's Disease Neuroimaging Initiative biomarkers as quantitative phenotypes: Genetics core aims, progress, and plans. Alzheimer's and

Dementia, 6, 265-273.

SCHEFF, S. W. AND PRICE, D. A. (2003). Synaptic pathology in Alzheimer's disease: a review of ultrastructural studies. Neurobiol Aging, 24, 1029-1046.

SCHIPPER, H. M., MAES, O. C., CHERTKOW, H. M. AND WANG, E. (2007). MicroRNA expression in Alzheimer blood mononuclear cells. Gene Regul Syst Bio, 1, 263-274.

SCHONROCK, N., HUMPHREYS, D. T., PREISS, T. AND GOTZ, J. (2011). Target Gene Repression Mediated by miRNAs miR-181c and miR-9 Both of Which Are Down- regulated by Amyloid-beta. J Mol Neurosci.

SCHONROCK, N., et al. (2010). Neuronal microRNA deregulation in response to Alzheimer's disease amyloid-beta. PLoS ONE, 5, e11070.

SCHONROCK, N., MATAMALES, M., ITTNER, L. M. AND GÖTZ, J. (2012). MicroRNA networks surrounding APP and amyloid- metabolism — Implications for Alzheimer's disease. Experimental Neurology, 235, 447-454.

SCHUCK, F., WOLF, D., FELLGIEBEL, A. AND ENDRES, K. (2016). Increase of alpha- Secretase ADAM10 in Platelets Along Cognitively Healthy Aging. J Alzheimers Dis, 6, 6. SEALS, D. F. AND COURTNEIDGE, S. A. (2003). The ADAMs family of metalloproteases:

Referências Bibliográficas

SERPENTE, M., et al. (2011). Role of OLR1 and Its Regulating hsa-miR369-3p in Alzheimer's Disease: Genetics and Expression Analysis. Journal of Alzheimer's Disease,

26, 787-793.

SHEINERMAN, K. S., TSIVINSKY, V. G., CRAWFORD, F., MULLAN, M. J., ABDULLAH, L. AND UMANSKY, S. R. (2012). Plasma microRNA biomarkers for detection of mild cognitive impairment. Aging, 4, 590-605.

SHUKLA, M., et al. (2015). Melatonin stimulates the nonamyloidogenic processing of betaAPP through the positive transcriptional regulation of ADAM10 and ADAM17. J

Pineal Res, 58, 151-165.

SMITH-VIKOS, T. AND SLACK, F. J. (2013). MicroRNAs circulate around Alzheimer's disease. Genome Biol, 14.

TAKEDA, S., SATO, N., RAKUGI, H. AND MORISHITA, R. (2010). Plasma [small beta]- amyloid as potential biomarker of Alzheimer disease: possibility of diagnostic tool for Alzheimer disease. Molecular BioSystems, 6.

TAKEUCHI, M., et al. (2007). Diagnostic utility of serum or cerebrospinal fluid levels of toxic advanced glycation end-products (TAGE) in early detection of Alzheimer's disease.

Med. Hypotheses, 69.

TANG, B. L. AND KUMAR, R. (2008). Biomarkers of mild cognitive impairment and Alzheimer's disease. Ann Acad Med Singapore, 37, 406-410.

THEENDAKARA, V., et al. (2013). Neuroprotective Sirtuin ratio reversed by ApoE4.

Proceedings of the National Academy of Sciences of the United States of America, 110,

18303-18308.

TIPPMANN, F., HUNDT, J., SCHNEIDER, A., ENDRES, K. AND FAHRENHOLZ, F. (2009). Up-regulation of the alpha-secretase ADAM10 by retinoic acid receptors and acitretin. Faseb J, 23, 1643-1654.

TROJANOWSKI, J. Q., et al. (2010). Update on the biomarker core of the Alzheimer's Disease Neuroimaging Initiative subjects. Alzheimers Dement, 6, 230-238.

VAN TETERING, G., VAN DIEST, P., VERLAAN, I., VAN DER WALL, E., KOPAN, R. AND VOOIJS, M. (2009). Metalloprotease ADAM10 is required for Notch1 site 2 cleavage. J Biol Chem, 284, 31018-31027.

VAN WART, H. E. AND BIRKEDAL-HANSEN, H. (1990). The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proceedings of the National Academy of Sciences of the

United States of America, 87, 5578-5582.

VILARDO, E., BARBATO, C., CIOTTI, M., COGONI, C. AND RUBERTI, F. (2010). MicroRNA-101 regulates amyloid precursor protein expression in hippocampal neurons. J

Referências Bibliográficas

WANG, Y., LIANG, Y. AND LU, Q. (2008). MicroRNA epigenetic alterations: predicting biomarkers and therapeutic targets in human diseases. Clin Genet, 74, 307-315.

WEBER, S. AND SAFTIG, P. (2012). Ectodomain shedding and ADAMs in development.

Development, 139, 3693-3709.

WONG, E., MARETZKY, T., PELEG, Y., BLOBEL, C. P. AND SAGI, I. (2015). The Functional Maturation of A Disintegrin and Metalloproteinase (ADAM) 9, 10, and 17 Requires Processing at a Newly Identified Proprotein Convertase (PC) Cleavage Site. J

Biol Chem, 290, 12135-12146.

YAMAZAKI, K., MIZUI, Y. AND TANAKA, I. (1997). Radiation hybrid mapping of human ADAM10 gene to chromosome 15. Genomics, 45, 457-459.

ZHANG, Y. W., THOMPSON, R., ZHANG, H. AND XU, H. (2011). APP processing in Alzheimer's disease. Mol Brain, 4, 3.

ZHONG, Z., et al. (2007). Levels of beta-secretase (BACE1) in cerebrospinal fluid as a predictor of risk in mild cognitive impairment. Arch Gen Psychiatry, 64, 718-726.

Manuscritos

4. MANUSCRITOS

4.1 MANUSCRITO I – Publicado

aDepartment of Gerontolgy, gDepartment of Physiological Sciences, hDepartment of

Medicine, Federal University of São Carlos, São Carlos, SP, Brazil

bDepartment of Pharmacological and Biomolecular Sciences,University of Studies of Milan,

Milan, Italy

cDepartment of Clinical and Experimental Science, Neurology Unit, University of Brescia,

Brescia, Italy

dNetherlands Institute for Neuroscience - an Institute of the Royal Netherlands Academy of

Arts and Sciences, Amsterdam, The Netherlands

eSwammerdam Institute for Life Sciences, Center for Neuroscience, University of

Amsterdam, Amsterdam, The Netherlands

fDepartment of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical

Center Utrecht, The Netherlands

* These authors equally contributed to this work

Corresponding Author: Márcia Regina Cominetti, Departamento de Gerontologia,

Universidade Federal de São Carlos, Rodovia Washington Luís, Km 235, São Carlos, SP, 13565-905, Brazil, phone: +55 16 3306 6663, fax +55 16 3351 9628. E-mail:

mcominetti@ufscar.br

Manuscritos

Abstract

ADAM10 is a potential biomarker for Alzheimer´s disease (AD). ADAM10 protein levels are reduced in platelets of AD patients. The aim was to verify the total blood and platelet ADAM10 gene expression in AD patients and to compare with mild cognitive impartment (MCI) and healthy subjects. No significant differences in ADAM10 gene expression were observed. Therefore, the decrease of ADAM10 protein in platelets of AD patients is not caused by a reduction in ADAM10 mRNA. Further studies must be performed to investigate other pathways in the down regulation of ADAM10 protein.

Manuscritos

1. Introduction

The pathological features in Alzheimer’s disease (AD) brain include cortical atrophy, predominantly in the medial temporal lobe and, microscopically, extensive neuronal loss and abnormal extra and intracellular fibrillar deposits, termed senile plaques and neurofibrillary tangles, respectively (Hohsfield and Humpel, 2015). The senile plaques appear from excessive deposition and subsequent aggregation of -amyloid peptide (A ) in the brain. AD pathogenesis is multifaceted and difficult to pinpoint, however genetic and cell biology studies led to the amyloid hypothesis, which posits that A plays a pivotal role in AD pathogenesis (Hardy and Selkoe, 2002). A derives from the concerted action of BACE1 ( - secretase) and the -secretase complex on the Amyloid Precursor Protein (APP) (Lichtenthaler, 2011). In healthy subjects the predominant route of APP processing consists of successive cleavages by and -secretases. In the non-amyloidogenic pathway, APP is cleaved by -secretases between lysine16 and leucine17 in the middle of A region, thus releasing sAPP - a structure with neurotrophic and neuroprotective functions (Meziane et al., 1998, Stein et al., 2004) retaining the C83 residue in the membrane. The following cleavage of C83 by -secretase releases the p3 - which is supposed to be beneficial, and is not found in amyloid plaques - and starts at position A 17 (A 17-40 and A 17-42), thereby inhibiting amyloidogenic A production (Morishima-Kawashima and Ihara, 2002).

Several enzymes in the ‘‘a disintegrin and metalloprotease’’ (ADAM) family, including ADAM9, ADAM10, and ADAM17, have -secretase activity in vitro, although recent studies have demonstrated that ADAM10 is the major -secretase that catalyzes APP ectodomain shedding in the brain (Kuhn et al., 2010, Jorissen et al., 2010). Moreover, it has been demonstrated that ADAM10 is a susceptibility gene of late onset AD (LOAD), the most common form of the disease. Indeed, two rare highly penetrant nonsynonymous mutations associated with LOAD have been identified in ADAM10 prodomain (Kim et al., 2009).

In previous studies we have reported a marked reduction in ADAM10 platelet protein levels in CDR (Clinical Dementia Rating) subgroups compared to non-AD patients (Manzine et al., 2013, Colciaghi et al., 2002) and APP, BACE1 and ADAM10 alterations in platelets already in the very early stages of the disease in which dementia can be barely inferred by neuropsychological assessments (Colciaghi et al., 2004). Since ADAM10 is the most important -secretase involved in cleavage of APP, in this work we raise the following question: is ADAM10 protein reduction in blood of AD patients a consequence of a decrease in ADAM10 transcription? In order to answer this question, we assessed the expression of

Manuscritos

ADAM10 mRNA in total blood and in platelets of a selected population of AD, MCI and control subjects, using Reverse Transcription quantitative PCR (RT-qPCR).

2. Methods

2.1 Characteristics of the Subjects

Patients were recruited in reference (Public Center of Specialties) and counter- reference (Family Health Units) health services in Brazil and at the Department of Medical Sciences-Neurology (University of Brescia, School of Medicine - Italy). Subjects recruited for AD group were diagnosed with probable AD according to National Institute of Neurological Disorders and Stroke-Alzheimer Disease and Related Disorders Association (NINCS- ADRDA) criteria. For MCI group, subjects had CDR 0.5, MoCA (range 19-25), Pfeffer test without impairment and follow the Petersen criteria (Petersen, 2004). All participants underwent the exclusion criteria for head trauma, metabolic dysfunctions, haematological diseases, alcohol abuse, drug abuse, delirium, mood disorders, and treatment with medications affecting platelet functions, i.e., anticoagulants, antiplatelet drugs, serotoninergic agonists-

Documentos relacionados